1
|
Haydara T, Gabr M, Abofreikha M, Bahnasy A, Salama H, Elhendawy M, Elkadeem M, Abd-Elsalam S. The Effect of Stem Cell Transplantation Therapy for Post Viral Chronic Liver Cell Failure on Associated Type II Diabetes Mellitus: A Pilot Study. Endocr Metab Immune Disord Drug Targets 2021; 20:903-916. [PMID: 31789137 DOI: 10.2174/1871530319666191202125402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/23/2019] [Accepted: 11/07/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND It was observed that type II diabetes mellitus associated with chronic liver failure improved after stem cell transplantation. However, there were no adequate studies regarding this issue. The aim of this study was to evaluate the effect of stem cell transplantation on associated type II diabetes mellitus and on the liver function tests. METHODS This pilot study included 30 patients of post-hepatitis chronic liver failure who were classified into two groups: Group I included patients with chronic liver cell failure associated with type 2 diabetes. Group II included patients without type II diabetes. Autologous CD34+ and CD133+ stem cells were percutaneously infused into the portal vein. Responders (regarding the improvement of diabetes as well as improvement of liver condition) and non-responders were determined. Patients were followed up for one, three and six months after the intervention evaluating their three-hour glucose tolerance test, C- peptide (Fasting and postprandial), Child-Pugh score and performance score one month, three months, and six months after stem cell therapy. RESULTS Both synthetic and excretory functions of the liver were improved in 10 patients (66.66 %) of group I and in 12 patients (80 %) of group II. Significant improvement in the Oral Glucose Tolerance Test in the responders of both the groups was well defined from the 3rd month and this was comparable to changes in liver function tests and Child-Pugh score. CONCLUSION Successful stem cell therapy in chronic liver cell failure patients can improve but not cure the associating type 2 diabetes by improving insulin resistance.
Collapse
Affiliation(s)
- Tamer Haydara
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mostafa Gabr
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Abofreikha
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Abeer Bahnasy
- Department of Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hosny Salama
- Department of Tropical Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohammed Elhendawy
- Department of Tropical Medicine and Infectious Diseases, faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mahmoud Elkadeem
- Department of Tropical Medicine and Infectious Diseases, faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sherief Abd-Elsalam
- Department of Tropical Medicine and Infectious Diseases, faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Greco EA, Lenzi A, Migliaccio S. The pathophysiological basis of bone tissue alterations associated with eating disorders. Horm Mol Biol Clin Investig 2016; 28:121-132. [DOI: 10.1515/hmbci-2016-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
Abstract
AbstractAnorexia nervosa (AN) and obesity are two major eating disorders present nowadays in Western countries. They are both characterized by striking body composition variations and hormonal alterations, which impact on skeletal metabolism, inducing bone tissue modifications and, thus, often cause an increased risk for fractures. AN and obesity are characterized by a severe reduction in fat mass and a high expression of it, respectively, and in both conditions hormones secreted or modulated by body fat content are important determinants of low bone density, impaired bone structure and reduced bone strength. In addition, in both AN and obesity, increased marrow adiposity, which correlates with low bone density, has been observed. This review will discuss the pathophysiological basis of bone alterations associated with AN and obesity, conditions of extreme energy deficiency and excess, respectively.
Collapse
|
3
|
Abstract
During the last decades, obesity and osteoporosis have become important global health problems, and the belief that obesity is protective against osteoporosis has recently come into question. In fact, some recent epidemiologic and clinical studies have shown that a high level of fat mass might be a risk factor for osteoporosis and fragility fractures. Several potential mechanisms have been proposed to explain the complex relationship between adipose tissue and bone. Indeed, adipose tissue secretes various molecules, named adipokines, which are thought to have effects on metabolic, skeletal and cardiovascular systems. Moreover, fat tissue is one of the major sources of aromatase, an enzyme that synthesizes estrogens from androgen precursors, hormones that play a pivotal role in the maintenance of skeletal homeostasis, protecting against osteoporosis. Moreover, bone cells express several specific hormone receptors and recent observations have shown that bone-derived factors, such as osteocalcin and osteopontin, affect body weight control and glucose homeostasis. Thus, the skeleton is considered an endocrine target organ and an endocrine organ itself, likely influencing other organs as well. Finally, adipocytes and osteoblasts originate from a common progenitor, a pluripotential mesenchymal stem cell, which has an equal propensity for differentiation into adipocytes or osteoblasts (or other lines) under the influence of several cell-derived transcription factors. This review will highlight recent insights into the relationship between fat and bone, evaluating both potential positive and negative influences between adipose and bone tissue. It will also focus on the hypothesis that osteoporosis might be considered the obesity of bone.
Collapse
Affiliation(s)
- Emanuela A. Greco
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Nutrition, ‘Sapienza’ University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Nutrition, ‘Sapienza’ University of Rome, Rome, Italy
| | - Silvia Migliaccio
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, Section of Health Sciences, ‘Foro Italico’ University of Rome, Largo Lauro De Bosis 15, 00195 Rome, Italy
| |
Collapse
|
4
|
Zhong L, Gou J, Deng N, Shen H, He T, Zhang BQ. Three-dimensional co-culture of hepatic progenitor cells and mesenchymal stem cells in vitro and in vivo. Microsc Res Tech 2015; 78:688-96. [PMID: 26031767 DOI: 10.1002/jemt.22526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/24/2015] [Accepted: 04/30/2015] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Here we co-cultured hepatic progenitor cells (HPCs) and mesenchymal stem cells (MSCs) to investigate whether the co-culture environments could increase hepatocytes form. METHODS Three-dimensional (3D) co-culture model of HPCs and MSCs was developed and morphological features of cells were continuously observed. Hepatocyte specific markers Pou5f1/Oct4, AFP, CK-18 and Alb were analyzed to confirm the differentiation of HPCs. The mRNA expression of CK-18 and Alb was analyzed by RT-PCR to investigate the influence of co-culture model to the terminal differentiation process of mature hepatocytes. The functional properties of hepatocyte-like cells were detected by continuously monitoring the albumin secretion using Gaussia luciferase assays. Scaffolds with HPCs and MSCs were implanted into nude mouse subcutaneously to set up the in vivo co-culture model. RESULTS Although two groups formed smooth spheroids and high expressed of CK-18 and Alb, hybrid spheroids had more regular structures and higher cell density. CK-18 and Alb mRNA were at a relatively higher expression level in co-culture system during the whole cultivation time (P < 0.05). Albumin secretion rates in the hybrid spheroids had been consistently higher than that in the mono-culture spheroids (P < 0.05). In vivo, the hepatocyte-like cells were consistent with the morphological features of mature hepatocytes and more well-differentiated hepatocyte-like cells were observed in the co-culture group. CONCLUSIONS HPCs and MSCs co-culture system is an efficient way to form well-differentiated hepatocyte-like cells, hence, may be helpful to the cell therapy of hepatic tissues and alleviate the problem of hepatocytes shortage.
Collapse
Affiliation(s)
- Li Zhong
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Juhua Gou
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Nian Deng
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Hao Shen
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Tongchuan He
- Department of Surgery, Molecular Oncology Laboratory, the University of Chicago Medical Center, Chicago, Illinois, 60637
| | - Bing-Qiang Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| |
Collapse
|
5
|
Gao B, Huang Q, Lin YS, Wei BY, Guo YS, Sun Z, Wang L, Fan J, Zhang HY, Han YH, Li XJ, Shi J, Liu J, Yang L, Luo ZJ. Dose-dependent effect of estrogen suppresses the osteo-adipogenic transdifferentiation of osteoblasts via canonical Wnt signaling pathway. PLoS One 2014; 9:e99137. [PMID: 24918446 PMCID: PMC4053448 DOI: 10.1371/journal.pone.0099137] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/11/2014] [Indexed: 12/26/2022] Open
Abstract
Fat infiltration within marrow cavity is one of multitudinous features of estrogen deficiency, which leads to a decline in bone formation functionality. The origin of this fat is unclear, but one possibility is that it is derived from osteoblasts, which transdifferentiate into adipocytes that produce bone marrow fat. We examined the dose-dependent effect of 17β-estradiol on the ability of MC3T3-E1 cells and murine bone marrow-derived mesenchymal stem cell (BMMSC)-derived osteoblasts to undergo osteo-adipogenic transdifferentiation. We found that 17β-estradiol significantly increased alkaline phosphatase activity (P<0.05); calcium deposition; and Alp, Col1a1, Runx2, and Ocn expression levels dose-dependently. By contrast, 17β-estradiol significantly decreased the number and size of lipid droplets, and Fabp4 and PPARγ expression levels during osteo-adipogenic transdifferentiation (P<0.05). Moreover, the expression levels of brown adipocyte markers (Myf5, Elovl3, and Cidea) and undifferentiated adipocyte markers (Dlk1, Gata2, and Wnt10b) were also affected by 17β-estradiol during osteo-adipogenic transdifferentiation. Western blotting and immunostaining further showed that canonical Wnt signaling can be activated by estrogen to exert its inhibitory effect of osteo-adipogenesis. This is the first study to demonstrate the dose-dependent effect of 17β-estradiol on the osteo-adipogenic transdifferentiation of MC3T3-E1 cells and BMMSCs likely via canonical Wnt signaling. In summary, our results indicate that osteo-adipogenic transdifferentiation modulated by canonical Wnt signaling pathway in bone metabolism may be a new explanation for the gradually increased bone marrow fat in estrogen-inefficient condition.
Collapse
Affiliation(s)
- Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Qiang Huang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yan-Shui Lin
- Department of Orthopaedics, First Affiliated Hospital, Chengdu Medical College, Chengdu, People’s Republic of China
| | - Bo-Yuan Wei
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yun-Shan Guo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Zhen Sun
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Long Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Jing Fan
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Hong-Yang Zhang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yue-Hu Han
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Xiao-Jie Li
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Jun Shi
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Jian Liu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
- * E-mail: (ZJL); (LY); (JL)
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
- * E-mail: (ZJL); (LY); (JL)
| | - Zhuo-Jing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
- * E-mail: (ZJL); (LY); (JL)
| |
Collapse
|
6
|
Tao H, Huang C, Yang JJ, Ma TT, Bian EB, Zhang L, Lv XW, Jin Y, Li J. MeCP2 controls the expression of RASAL1 in the hepatic fibrosis in rats. Toxicology 2011; 290:327-33. [PMID: 22056649 DOI: 10.1016/j.tox.2011.10.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/17/2011] [Accepted: 10/19/2011] [Indexed: 12/11/2022]
Abstract
Hepatic stellate cells (HSCs) activation is an essential event during liver fibrogenesis. A major pathway is the transition of HSCs into hepatic myofibroblasts. The methyl-CpG-binding protein MeCP2 which promotes repressed chromatin structure is selectively detected in myofibroblasts of diseased liver. Overexpression of this protein results in an increase of global methylation levels. Treatment of HSCs with DNA methylation inhibitor 5-aza-2'-deoxycytidine (5-azadC) blocks the cell proliferation. 5-azadC also prevents loss of Ras GTPase activating-like protein 1 (RASAL1) expression that occurs during HSCs proliferation. To further explore the underlying molecular mechanisms, we hypothesized that this perpetuation of fibrogenesis was caused by DNA methylation. Results demonstrated that hypermethylation of RASAL1 is associated with the perpetuation of fibroblast activation and fibrogenesis in the liver. knockdown of MeCP2 using siRNA technique increased RASAL1 in both mRNA and protein level in myofibroblasts. These studies demonstrated that MeCP2 and DNA methylation may provide molecular mechanisms for perpetuated fibroblast activation and fibrogenesis in the liver.
Collapse
Affiliation(s)
- Hui Tao
- School of pharmacy, Anhui key laboratory of bioactivity of natural products, Anhui Medical University, Hefei 230032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
Cell therapy is based on the replacement of damaged cells in order to restore injured tissues. The first consideration is that an abundant source of cells is needed; second, these cells should be immunologically compatible with the guest and third, there should be no real threat of these cells undergoing malignant transformation in the future. Given these requirements, already differentiated adult cells or adult stem cells obtained from the body of the patient appear to be the ideal candidates to meet all of these demands. The utilization of somatic cells also avoids numerous ethical and political drawbacks and concerns. Transdifferentiation is the phenomenon by which an adult differentiated cell switches to another differentiated cell. This paper reviews the importance of transdifferentiation, discussing the cells that are suitable for this process and the methods currently employed to induce the change in cell type.
Collapse
|
8
|
Wallace K, Long Q, Fairhall EA, Charlton KA, Wright MC. Serine/threonine protein kinase SGK1 in glucocorticoid-dependent transdifferentiation of pancreatic acinar cells to hepatocytes. J Cell Sci 2011; 124:405-13. [PMID: 21224398 DOI: 10.1242/jcs.077503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Elevated glucocorticoid levels result in the transdifferentiation of pancreatic acinar cells into hepatocytes through a process that requires a transient repression of WNT signalling upstream of the induction of C/EBP-β. However, the mechanism by which glucocorticoid interacts with WNT signalling is unknown. A screen of microarray data showed that the serine/threonine protein kinase SGK1 (serum- and glucocorticoid-regulated kinase 1) was markedly induced in the model B-13 pancreatic rat acinar cell line after glucocorticoid treatment (which converts them into hepatocyte-like 'B-13/H' cells) and this was confirmed at the level of mRNA (notably an alternatively transcribed SGK1C form) and protein. Knockdown of SGK1 using an siRNA designed to target all variant transcripts inhibited glucocorticoid-dependent transdifferentiation, whereas overexpression of the human C isoform (and also the human SGK1F isoform, for which no orthologue in the rat has been identified) alone - but not the wild-type A form - inhibited distal WNT signalling Tcf/Lef transcription factor activity, and converted B-13 cells into B-13/H cells. These effects were lost when the kinase functions of SGK1C and SGK1F were mutated. Inhibition of SGK1 kinase activity also inhibited glucocorticoid-dependent transdifferentiation. Expression of SGK1C and SGK1F resulted in the appearance of phosphorylated β-catenin, and recombinant SGK1 was shown to directly phosphorylate purified β-catenin in vitro in an ATP-dependent reaction. These data therefore demonstrate a crucial role for SGK1 induction in B-13 cell transdifferentiation to B-13/H hepatocytes and suggest that direct phosphorylation of β-catenin by SGK1C represents the mechanism of crosstalk between glucocorticoid and WNT signalling pathways.
Collapse
Affiliation(s)
- Karen Wallace
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE24HH, UK
| | | | | | | | | |
Collapse
|
9
|
Abstract
Lens regeneration among vertebrates is basically restricted to some amphibians. The most notable cases are the ones that occur in premetamorphic frogs and in adult newts. Frogs and newts regenerate their lens in very different ways. In frogs the lens is regenerated by transdifferentiation of the cornea and is limited only to a time before metamorphosis. On the other hand, regeneration in newts is mediated by transdifferentiation of the pigment epithelial cells of the dorsal iris and is possible in adult animals as well. Thus, the study of both systems could provide important information about the process. Molecular tools have been developed in frogs and recently also in newts. Thus, the process has been studied at the molecular and cellular levels. A synthesis describing both systems was long due. In this review we describe the process in both Xenopus and the newt. The known molecular mechanisms are described and compared.
Collapse
Affiliation(s)
- Jonathan J Henry
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA.
| | | |
Collapse
|
10
|
Wallace K, Marek CJ, Hoppler S, Wright MC. Glucocorticoid-dependent transdifferentiation of pancreatic progenitor cells into hepatocytes is dependent on transient suppression of WNT signalling. J Cell Sci 2010; 123:2103-10. [DOI: 10.1242/jcs.070722] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Developmentally, the pancreas and liver are closely related and pathological conditions – including elevated glucocorticoid levels – result in the appearance of hepatocytes in the pancreas. The role of the WNT signalling pathway in this process has been examined in the model transdifferentiating pancreatic acinar AR42J-B-13 (B-13) cell. Glucocorticoid treatment resulted in a transient loss of constitutive WNT3a expression, phosphorylation and depletion of β-catenin, loss of β-catenin nuclear localisation, and significant reductions in T-cell factor/lymphoid enhancer factor (Tcf/Lef) transcriptional activity before overt changes in phenotype into hepatocyte-like (B-13/H) cells. A return to higher Tcf/Lef transcriptional activity correlated with the re-expression of WNT3a in B-13/H cells. β-catenin knock down alone substituted for and enhanced glucocorticoid-dependent transdifferentiation. Overexpression of a mutant β-catenin (pt-Xβ-cat) protein that blocked glucocorticoid-dependent suppression of Tcf/Lef activity resulted in inhibition of transdifferentiation. A small-molecule activator of Tcf/Lef transcription factors blocked glucocorticoid-dependent effects, as observed with pt-Xβ-cat expression. Quercetin – a Tcf/Lef inhibitor – did not promote transdifferentiation into B-13/H cells, but did potentiate glucocorticoid-mediated transdifferentiation. These data demonstrate that the transdifferentiation of B-13 cells into hepatocyte-like cells in response to glucocorticoid was dependent on the repression of constitutively active WNT signalling.
Collapse
Affiliation(s)
- Karen Wallace
- Institute of Cellular Medicine, Level 2 Leech Building, Medical School, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
| | - Carylyn J. Marek
- Institute of Cellular Medicine, Level 2 Leech Building, Medical School, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Matthew C. Wright
- Institute of Cellular Medicine, Level 2 Leech Building, Medical School, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
| |
Collapse
|
11
|
Beck CW, Izpisúa Belmonte JC, Christen B. Beyond early development: Xenopus as an emerging model for the study of regenerative mechanisms. Dev Dyn 2009; 238:1226-48. [PMID: 19280606 DOI: 10.1002/dvdy.21890] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
While Xenopus is a well-known model system for early vertebrate development, in recent years, it has also emerged as a leading model for regeneration research. As an anuran amphibian, Xenopus laevis can regenerate the larval tail and limb by means of the formation of a proliferating blastema, the lens of the eye by transdifferentiation of nearby tissues, and also exhibits a partial regeneration of the postmetamorphic froglet forelimb. With the availability of inducible transgenic techniques for Xenopus, recent experiments are beginning to address the functional role of genes in the process of regeneration. The use of soluble inhibitors has also been very successful in this model. Using the more traditional advantages of Xenopus, others are providing important lineage data on the origin of the cells that make up the tissues of the regenerate. Finally, transcriptome analyses of regenerating tissues seek to identify the genes and cellular processes that enable successful regeneration. Developmental Dynamics 238:1226-1248, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Caroline W Beck
- Department of Zoology and Genetics Otago, University of Otago, New Zealand.
| | | | | |
Collapse
|
12
|
In vitro transdifferentiation of human hepatoma cells into pancreatic-like cells. Methods Mol Biol 2009. [PMID: 19504247 DOI: 10.1007/978-1-59745-448-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Transdifferentiation is defined as an irreversible switch in postnatal life of one differentiated cell to another. Transdifferentiation from different cellular origins into pancreatic-like beta-cells is of clinical significance since this approach may offer a potential cure for diabetes. In order to achieve this goal, the liver is considered as a suitable candidate due to its close developmental relationship to the pancreas, its large size and a well-documented regenerative capacity that could provide enough original tissues to initiate the transdifferentiation procedure. In this chapter, we describe a protocol to overexpress Pdx1, a master regulator essential for pancreas development in the cultured human liver cell line, HepG2.
Collapse
|
13
|
Denner L, Urban RJ. Critical issues for engineering cord blood stem cells to produce insulin. Expert Opin Biol Ther 2008; 8:1251-4. [PMID: 18694347 DOI: 10.1517/14712598.8.9.1251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND/OBJECTIVES The objectives of using cord blood stem cells for treating type 1 diabetes are simple in principle yet complex in biological and molecular mechanisms. These are defined by the complexity of the insulin-producing unit of the pancreas, the islet. Islets are composed of various cell types that arise from diverse lineages and communicate by hormones, growth factors and small-molecule mediators. These processes are regulated by integration of signal transduction pathways. While advances have been made to engineer umbilical cord blood stem cells to produce insulin, these studies only illuminate the potential of such cells to fulfil a necessary, but not sufficient, requirement for transplantation. RESULTS/CONCLUSIONS The challenges ahead demand detailed understanding of molecular mechanisms to move from an opportunistic, phenotypic approach to transplantation and amelioration of blood glucose, to an orderly and logical approach to a biologically and medically meaningful solution. The issues include expansion to generate large numbers of cells, self-renewal to regulate the destiny of cord blood stem cells to repopulate the hematopoietic system, and multipotency of stem cells to generate the distinct cell types of an islet.
Collapse
|
14
|
Li HY, Zhou XF. Potential conversion of adult clavicle-derived chondrocytes into neural lineage cells in vitro. J Cell Physiol 2008; 214:630-44. [PMID: 17786944 DOI: 10.1002/jcp.21251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neural stem cells (NSC) can be isolated from a variety of adult tissues and become a valuable cell source for the repair of peripheral and central nervous diseases. However, their origin and identity remain controversial because of possible de-differentiation/trans-differentiation or contaminations by hematopoietic stem cells (HSCs) or mesenchymal stem cells (MSCs). We hypothesize that the commonly used NSC culture medium can induce committed cartilage chondrocytes to de-differentiate and/or trans-differentiate into neural cell lineages. Using a biological isolation and purification method with explants culture, we here show that adult rat clavicle cartilage chondrocytes migrate out from tissue blocks, form sphere-like structures, possess the capability of self-renewal, express nestin and p75NTR, markers for neural crest progenitors, and differentiate into neurons, glia, and smooth muscle cells. Comparing with adult cartilage, the spherical-forming neural crest cell-like cells downregulate the chondrocytic marker genes, including collagen II, collagen X, and sox9, as well as neural-lineage repressors/silencers REST and coREST, but upregulate a set of well-defined genes related to neural crest cells and pro-neural potential. Nerve growth factor (NGF) and glial growth factor (GGF) increase glial and neuronal differentiation, respectively. These results suggest that chondrocytes derived from adult clavicle cartilage can become neural crest stem-like cells and acquire neuronal phenotypes in vitro. The possible de-differentiation/trans-differentiation mechanisms underlying the conversion were discussed.
Collapse
Affiliation(s)
- Hong-Yun Li
- Department of Human Physiology, School of Medicine, Flinders University of South Australia, Adelaide, South Australia, Australia
| | | |
Collapse
|
15
|
Qihao Z, Xigu C, Guanghui C, Weiwei Z. Spheroid formation and differentiation into hepatocyte-like cells of rat mesenchymal stem cell induced by co-culture with liver cells. DNA Cell Biol 2007; 26:497-503. [PMID: 17630854 DOI: 10.1089/dna.2006.0562] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from bone marrow have been shown to differentiate into hepatocytes, which would be an ideal resource for transplantation or artificial liver devices. Here we investigated the efficiency of co-culture system consisting of rat MSCs and adult liver cells to induce differentiation of MSCs into hepatocyte-like cells. Marked MSCs were either co-cultured with freshly isolated liver cells or treated with hepatocyte growth factor (HGF) for 21 days. In co-culture systems, MSCs formed spheroids of round-shaped cells while keeping normal proliferation and viability, strongly expressed albumin, alpha-fetoprotein, and cytokeratin-18 in mRNA and protein level from day 3 to 21. As a control, MSCs treated with HGF showed weak gene expressions in day 14 and had a few cells of protein staining in day 21. These results indicate that the co-culture microenvironment plays a decisive role for the hepatic differentiation of MSCs, and it is more efficient than HGF treatment. Insights gained from this study will be helpful to design optimal culture systems for the hepatic differentiation of human MSCs and the hepatic function maintenance of hepatocytes in vitro.
Collapse
Affiliation(s)
- Zhang Qihao
- Center of Experimental Animals, Sun Yat-Sen University, Guangzhou, China
| | | | | | | |
Collapse
|
16
|
Pouton CW, Haynes JM. Embryonic stem cells as a source of models for drug discovery. Nat Rev Drug Discov 2007; 6:605-16. [PMID: 17667955 DOI: 10.1038/nrd2194] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) will become a source of models for a wide range of adult differentiated cells, providing that reliable protocols for directed differentiation can be established. Stem-cell technology has the potential to revolutionize drug discovery, making models available for primary screens, secondary pharmacology, safety pharmacology, metabolic profiling and toxicity evaluation. Models of differentiated cells that are derived from mouse ESCs are already in use in drug discovery, and are beginning to find uses in high-throughput screens. Before analogous human models can be obtained in adequate numbers, reliable methods for the expansion of human ESC cultures will be needed. For applications in drug discovery, involving either species, protocols for directed differentiation will need to be robust and affordable. Here, we explore current challenges and future opportunities in relation to the use of stem-cell technology in drug discovery, and address the use of both mouse and human models.
Collapse
Affiliation(s)
- Colin W Pouton
- Department of Pharmaceutical Biology, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Melbourne, Australia.
| | | |
Collapse
|
17
|
Denner L, Bodenburg Y, Zhao JG, Howe M, Cappo J, Tilton RG, Copland JA, Forraz N, McGuckin C, Urban R. Directed engineering of umbilical cord blood stem cells to produce C-peptide and insulin. Cell Prolif 2007; 40:367-80. [PMID: 17531081 PMCID: PMC6496474 DOI: 10.1111/j.1365-2184.2007.00439.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES In this study, we investigated the potential of umbilical cord blood stem cell lineages to produce C-peptide and insulin. MATERIALS AND METHODS Lineage negative, CD133+ and CD34+ cells were analyzed by flow cytometry to assess expression of cell division antigens. These lineages were expanded in culture and subjected to an established protocol to differentiate mouse embryonic stem cells (ESCs) toward the pancreatic phenotype. Phase contrast and fluorescence immunocytochemistry were used to characterize differentiation markers with particular emphasis on insulin and C-peptide. RESULTS All 3 lineages expressed SSEA-4, a marker previously reported to be restricted to the ESC compartment. Phase contrast microscopy showed all three lineages recapitulated the treatment-dependent morphological changes of ESCs as well as the temporally restricted expression of nestin and vimentin during differentiation. After engineering, each isolate contained both C-peptide and insulin, a result also obtained following a much shorter protocol for ESCs. CONCLUSIONS Since C-peptide can only be derived from de novo synthesis and processing of pre-proinsulin mRNA and protein, we conclude that these results are the first demonstration that human umbilical cord blood-derived stem cells can be engineered to engage in de novo synthesis of insulin.
Collapse
Affiliation(s)
- L Denner
- Stark Diabetes Center and McCoy Diabetes Mass Spectrometry Research Laboratory, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555-1060, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mann J, Oakley F, Akiboye F, Elsharkawy A, Thorne AW, Mann DA. Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: implications for wound healing and fibrogenesis. Cell Death Differ 2007; 14:275-85. [PMID: 16763620 DOI: 10.1038/sj.cdd.4401979] [Citation(s) in RCA: 192] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Myofibroblasts are critical cellular elements of wound healing generated at sites of injury by transdifferentiation of resident cells. A paradigm for this process is conversion of hepatic stellate cells (HSC) into hepatic myofibroblasts. Treatment of HSC with DNA methylation inhibitor 5-aza-2'-deoxycytidine (5-azadC) blocked transdifferentiation. 5-azadC also prevented loss of IkappaBalpha and PPARgamma expression that occurs during transdifferentiation to allow acquisition of proinflammatory and profibrogenic characteristics. ChIP analysis revealed IkappaBalpha promoter is associated with transcriptionally repressed chromatin that converts to an active state with 5-azadC treatment. The methyl-CpG-binding protein MeCP2 which promotes repressed chromatin structure is selectively detected in myofibroblasts of diseased liver. siRNA knockdown of MeCP2 elevated IkappaBalpha promoter activity, mRNA and protein expression in myofibroblasts. MeCP2 interacts with IkappaBalpha promoter via a methyl-CpG-dependent mechanism and recruitment into a CBF1 corepression complex. We conclude that MeCP2 and DNA methylation exert epigenetic control over hepatic wound healing and fibrogenesis.
Collapse
Affiliation(s)
- J Mann
- Liver Group, Division of Infection, Inflammation and Repair, University of Southampton, Southampton General Hospital, Tremona Road, Southampton SO166YD, UK
| | | | | | | | | | | |
Collapse
|
19
|
Roh J, Cho EA, Seong I, Limb JK, Lee S, Han SJ, Kim J. Down-regulation of Sox10 with specific small interfering RNA promotes transdifferentiation of Schwannoma cells into myofibroblasts. Differentiation 2006; 74:542-51. [PMID: 17177851 DOI: 10.1111/j.1432-0436.2006.00084.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neural crest-derived cells are noted for the long lasting plasticity during lineage commitment process and the potential for transdifferentiation into other neural crest derivatives. Schwann cells in particular have been reported to transdifferentiate into melanocytes and myofibroblasts. Detailed studies of transdifferentiation at the molecular level have been hampered by difficulty in isolating sufficient quantity of primary cells or cellular materials. Here, we describe a robust in vitro system in which Schwannoma cells undergo an apparent transdifferentiation into myofibroblasts. Importantly, we induce the transdifferentiation by down-regulating a single transcription factor, Sox10, thereby identifying a key molecular event in this process. Myofibroblasts thus generated showed carbachol-stimulated contraction and calcium transients and express several established myofibroblast-specific genes. These results suggest that generating desired cell types based on "knock-down" of critical genes may be a viable strategy.
Collapse
Affiliation(s)
- Jiwon Roh
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Sudaemun-gu, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Ong SY, Dai H, Leong KW. Inducing hepatic differentiation of human mesenchymal stem cells in pellet culture. Biomaterials 2006; 27:4087-97. [PMID: 16616366 DOI: 10.1016/j.biomaterials.2006.03.022] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Accepted: 03/15/2006] [Indexed: 11/25/2022]
Abstract
Extensive cell-cell or cell-matrix interaction in three-dimensional (3D) culture is important for the maintenance of adult hepatocyte function and the maturation of hepatic progenitors. However, although there is significant interest in inducing the transdifferentiation of adult stem cells into the hepatic lineage, very few studies have been conducted in a 3D culture configuration. The aim of this study is to investigate the differentiation of mesenchymal stem cells (MSC) into hepatocytes in a pellet configuration, with or without the presence of small intestinal submucosa (SIS). After 4 weeks of differentiation with growth factors bFGF, HGF, and OsM, we obtained hepatocyte-like cells that expressed a subset of hepatic genes, secreted albumin and urea, stored glycogen, and showed inducible CYP3A4 mRNA levels. When these cells were implanted into livers of hepatectomized rats, they secreted human albumin into the bloodstream. The hepatic differentiation of MSC was faster in cell pellets without SIS. The plausible explanations for this finding may be related to the mass transport issues of the two different pellets and the role of cell-cell contact over cell-matrix interactions. The findings of this study should help in the design of optimal culture configurations for efficient hepatic differentiation of adult stem cells.
Collapse
Affiliation(s)
- Shin-Yeu Ong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
21
|
Chen YK, Zhao XX, Li JG, Lang S, Wang YM. Ductular proliferation in liver tissues with severe chronic hepatitis B: An immunohistochemical study. World J Gastroenterol 2006; 12:1443-6. [PMID: 16552818 PMCID: PMC4124327 DOI: 10.3748/wjg.v12.i9.1443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To clarify the pathogenesis of ductular proliferation and its possible association with oval cell activation and hepatocyte regeneration.
METHODS: Immunohistochemical staining and image analysis of the ductular structures in the liver tissues from 11 patients with severe chronic hepatitis B and 2 healthy individuals were performed. The liver specimens were sectioned serially, and then cytokeratin 8 (CK8), CK19, OV6, proliferating cell nuclear antigens (PCNA), glutathione-S-transferase (GST), α-fetal protein (AFP) and albumin were stained immunohistochemically.
RESULTS: Typical and atypical types of ductular proliferation were observed in the portal tracts of the liver tissues in all 11 patients. The proliferating ductular cells were positive for CK8, CK19, OV6 and PCNA staining. Some atypical ductular cells displayed the morphological and immunohistochemical characteristics of hepatic oval cells. Some small hepatocyte-like cells were between hepatic oval cells and mature hepatocytes morphometrically and immunohistochemically.
CONCLUSION: The proliferating ductules in the liver of patients with severe chronic liver disease may have different origins. Some atypical ductular cells are actually activated hepatic oval cells. Atypical ductular proliferation is related to hepatocyte regeneration and small hepatocyte-like cells may be intermediate transient cells between hepatic oval cells and mature hepatocytes.
Collapse
Affiliation(s)
- Yao-Kai Chen
- Institute of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | | | | | | | | |
Collapse
|
22
|
Kindler V. Postnatal stem cell survival: does the niche, a rare harbor where to resist the ebb tide of differentiation, also provide lineage-specific instructions? J Leukoc Biol 2006; 78:836-44. [PMID: 16199730 DOI: 10.1189/jlb.0505272] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Postnatal stem cells regulate the homeostasis of the majority of our tissues. They continuously generate new progenitors and mature, functional cells to replace old cells, which cannot assume the tissue function anymore and are eliminated. Blood, skin, gut mucosa, muscle, cartilage, nerves, cornea, retina, liver, and many other structures are regulated by stem cells. As a result of their ability to produce large numbers of functionally mature cells, postnatal stem cells represent a promising tool for regenerative therapy. Indeed, unmanipulated stem cells or their progeny amplified in vitro are already used in some clinical applications to restore the function of injured or genetically deficient tissues. However, despite our cumulating understanding concerning postnatal stem cells, many aspects of their functionality remain unclear. For instance, in most tissues, we cannot reliably define the phenotype of the postnatal stem cells sustaining its survival. We do not know to which extent the environment surrounding the stem cell-the niche-which is a key actor insuring stem cell self-maintenance, is also implicated in the maintenance of stem cell lineage specificity. Moreover, we have to clarify whether postnatal stem cells are capable of undertaking "transdifferentiation", that is, the conversion of one cell type into another under physiological conditions. Answering these questions should help us to draw a more accurate picture of postnatal stem cell biology and should lead to the design of safe, effective therapies.
Collapse
Affiliation(s)
- Vincent Kindler
- Geneva University Hospital, 25, Micheli-du-Crest, 1211 Geneva 14, Switzerland.
| |
Collapse
|
23
|
Abstract
The authors reviewed 77 published reports available before August 1, 2005 that examined the ability of hematopoietic cells to generate hepatocytes in the liver. A list of these publications and a synopsis of each are available on-line. We interpret the evidence provided by this data set to suggest that one or more types of hematopoietic cells may rarely acquire the hepatocyte phenotype in the liver (frequency < or =10(-4)), although the nature of the hematopoietic cells involved and the mechanisms responsible for acquisition of a hepatocyte phenotype are still controversial. Hematopoietic stem cells do not appear to be direct precursors of hepatocytes, which, instead, can be generated from cells of the macrophage-monocyte lineage. Fusion between hepatocytes and transplanted hematopoietic cells has been substantiated as a mechanism by which hepatocytes that carry a bone marrow tag are generated, but direct transdifferentiation of hematopoietic cells has not been demonstrated. In conclusion, hematopoietic cells contribute little to hepatocyte formation under either physiological or pathological conditions, although they may provide cytokines and growth factors that promote hepatocyte functions by paracrine mechanisms. Cells of the endodermal hepatocyte lineage are far more potent generators of hepatocytes than are hematopoietic cells.
Collapse
Affiliation(s)
- Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, MD 20892-4262, USA.
| | | |
Collapse
|
24
|
Monier B, Astier M, Sémériva M, Perrin L. Steroid-dependent modification of Hox function drives myocyte reprogramming in the Drosophila heart. Development 2005; 132:5283-93. [PMID: 16284119 DOI: 10.1242/dev.02091] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the Drosophila larval cardiac tube, aorta and heart differentiation are controlled by the Hox genes Ultrabithorax(Ubx) and abdominal A (abdA), respectively. There is evidence that the cardiac tube undergoes extensive morphological and functional changes during metamorphosis to form the adult organ, but both the origin of adult cardiac tube myocytes and the underlying genetic control have not been established. Using in vivo time-lapse analysis, we show that the adult fruit fly cardiac tube is formed during metamorphosis by the reprogramming of differentiated and already functional larval cardiomyocytes,without cell proliferation. We characterise the genetic control of the process, which is cell autonomously ensured by the modulation of Ubxexpression and AbdA activity. Larval aorta myocytes are remodelled to differentiate into the functional adult heart, in a process that requires the regulation of Ubx expression. Conversely, the shape, polarity,function and molecular characteristics of the surviving larval contractile heart myocytes are profoundly transformed as these cells are reprogrammed to form the adult terminal chamber. This process is mediated by the regulation of AbdA protein function, which is successively required within these persisting myocytes for the acquisition of both larval and adult differentiated states. Importantly, AbdA specificity is switched at metamorphosis to induce a novel genetic program that leads to differentiation of the terminal chamber. Finally, the steroid hormone ecdysone controls cardiac tube remodelling by impinging on both the regulation of Ubx expression and the modification of AbdA function. Our results shed light on the genetic control of one in vivo occurring remodelling process, which involves a steroid-dependent modification of Hox expression and function.
Collapse
Affiliation(s)
- Bruno Monier
- Laboratoire de Génétique et Physiologie du Développement, UMR 6545 CNRS-Université, Campus de Luminy, Case 907, 13288 Marseille Cedex 09, France
| | | | | | | |
Collapse
|